32
A comprehensive comparison of comparative RNA structure prediction approaches Paul P. Gardner *1 Robert Giegerich 2 1 Department of Evolutionary Biology, University of Copenhagen, Universitetsparken 15, 2100 Copenhagen Ø, Denmark 2 Faculty of Technology, University of Bielefeld, PO Box 10 01 31, 33501 Bielefeld, Germany Email: Paul P. Gardner * - [email protected]; Robert Giegerich - [email protected]; * Corresponding author Abstract Background: An increasing number of researchers have released novel RNA structure analysis and prediction algorithms for comparative approaches to structure prediction. Yet, independent benchmarking of these algorithms is rarely performed as is now common practice for protein-folding, gene-finding and multiple-sequence-alignment algorithms. Results: Here we evaluate a number of RNA folding algorithms upon reliable RNA data-sets and compare their relative performance. Conclusions: We conclude that comparative data can enhance structure prediction but structure prediction, algorithms vary widely in terms of both sensitivity and selectivity. Furthermore, we outline some directions for future research. Background Motivation RNA, once considered a passive carrier of genetic information, is now known to play a more active role in nature. Many recently discovered RNAs are catalytic, for example RNase P which is involved in tRNA maturation and the self-splicing introns involved in mRNA maturation [1]. In addition, there is evidence 1

A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

A comprehensive comparison of comparative RNA structureprediction approaches

Paul P. Gardner∗1 Robert Giegerich2

1Department of Evolutionary Biology, University of Copenhagen, Universitetsparken 15, 2100 Copenhagen Ø, Denmark2Faculty of Technology, University of Bielefeld, PO Box 10 01 31, 33501 Bielefeld, Germany

Email: Paul P. Gardner∗- [email protected]; Robert Giegerich - [email protected];

∗Corresponding author

Abstract

Background: An increasing number of researchers have released novel RNA structure analysis and prediction

algorithms for comparative approaches to structure prediction. Yet, independent benchmarking of these

algorithms is rarely performed as is now common practice for protein-folding, gene-finding and

multiple-sequence-alignment algorithms.

Results: Here we evaluate a number of RNA folding algorithms upon reliable RNA data-sets and compare their

relative performance.

Conclusions: We conclude that comparative data can enhance structure prediction but structure prediction,

algorithms vary widely in terms of both sensitivity and selectivity. Furthermore, we outline some directions for

future research.

BackgroundMotivation

RNA, once considered a passive carrier of genetic information, is now known to play a more active role in

nature. Many recently discovered RNAs are catalytic, for example RNase P which is involved in tRNA

maturation and the self-splicing introns involved in mRNA maturation [1]. In addition, there is evidence

1

Page 2: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

that RNA based organisms were an essential step in the evolution of modern DNA-protein based

organisms [2, 3]. The number of non-coding RNAs (ncRNA) in humans remains a mystery, but progress in

this direction suggests the number of ncRNAs produced is comparable to the number of proteins [4–6].

Surprisingly, the number of protein coding genes does not correlate with our concept of “organism

complexity”, hence it has been hypothesised that control of gene expression via a combination of

alternative splicing and non-coding RNAs are responsible for this, implying that the “Central Dogma”

(RNA is transcribed from DNA and translated into protein) at least in higher eukaryotes is woefully

inadequate [7, 8].

A fundamental tenet of biology is that a stable tertiary structure is essential for biological function. In the

case of RNA the secondary structure (the base-pair set for an RNA molecule) provides a scaffold for the

tertiary structure [9, 10]. Yet, the experimental determination of RNA structure remains difficult [11];

Researchers increasingly turn to computational methods. To date the most popular structure prediction

algorithm is the Minimum Free Energy (MFE) method for folding a single sequence, this has been

implemented by two packages: Mfold [12] and RNAfold [13]. However, there are several independent

reasons why the accuracy of MFE structure prediction is limited in practise (see discussion below).

Generally the best accuracy can be achieved by employing comparative methods [14]. This paper explores

the extent to which this statement is true, given the current state of the art, for automated methods.

There are currently three approaches to automated comparative RNA sequence analysis where the

comparative study is supported by available algorithms (see plans A, B, and C, figure ??). A researcher

following Plan A may align sequences using standard multiple sequence alignment tools (i.e. clustalw [15],

t-coffee [16], prrn [17],. . .), then use the signal provided by structure neutral mutations for the inference of

a consensus structure. Frequently the mutual-information measure is used for this [18–20]. Recently tools

have been developed that use a combination of MFE and a covariation-score [21,22] or probabilistic models

compiled from large reference data-sets [23, 24]. However, a multiple-sequence-alignment step assumes a

well conserved sequence. This is often not so with swiftly evolving ncRNA sequences, in this case incorrect

sequence alignments can destroy the covariation signal.

This has motivated Plan B, the use of the “Sankoff-Algorithm”, an algorithm designed for the

simultaneous alignment, folding and inference of a protosequence for a set of homologous structural RNA

sequences [25]. The recurrences combine sequence alignment and Nussinov (maximal pairing) folding [26].

The algorithm requires extreme computational resources (O(n6) in time, and O(n4) in space, where n is

the sequence length). Current implementations, Foldalign [27,28], Dynalign [29] and PMmulti [26], are

2

Page 3: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

restricted implementations of the Sankoff-algorithm which impose pragmatic limits on the size or shape of

substructures.

The final approach (Plan C) applies when no helpful level of sequence conservation is observed. We may

exclude the sequence alignment step, predict secondary structures for each sequence (or sub-group of

sequences) separately, and directly align the structures. Because of the nested branching nature of RNA

structures, these are adequately represented as trees. The concept of a similarity measurement via edit

operations, a standard procedure for string comparisons, has been generalised to trees [30–33]. Tree

comparison and tree alignment models have been proposed [34,35] and implemented [13,36–39]. The

crucial point in Plan C is the question whether the initial independent folding produces at least some

structures that align well and hence give clues as to the underlying consensus structure – when one exists.

An increasing number of researchers have recently released novel RNA structure analysis and prediction

algorithms [22,23,37,40–43]. Yet few algorithms are tested upon standardised example data-sets, and often

they are not compared with algorithms of the same pedigree. Algorithm evaluation is a regular event for

protein structure prediction groups [44–47], gene-prediction [48–50] and multiple sequence

alignments [51–54]. The concrete goals of this study are as follows: Based on reliable data-sets, we evaluate:

• the viability of plan A, B, or C given tools available today, and

• the relative performance of the tools used within each plan.

We shall explicitly not evaluate computational efficiency, which (by necessity) differs widely between the

tools. We also do not evaluate user friendliness (such as ease of installation and convenience of input or

output formats, etc.) except for some remarks in the discussion section. Data-sets, documentation and

relevant scripts are freely available from www.binf.ku.dk/users/pgardner/bralibase/.

Structural alignments and consensus structures

RNA secondary structure inference is the prediction of base-pairs which form the in vivo structure, given

only the sequence of bases. Three general considerations apply: (1) The in vivo structure is not only

predetermined by the primary structure, but also by cellular components such as chaperones, base

modifications, and even by the transcriptional process itself. There are currently no computational tools

available that assess these effects. (2) There are ‘ribo-switches’, whereby two or more functional structures

exist for a given sequence [55–57]. Such cases will fool all the tools studied here, because asking for a single

consensus structure is simply the wrong question. On the other hand, the potential of conformational

3

Page 4: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

switching can be reliably detected [58–60]. (3) Structures may occasionally contain pseudo-knots, which

are ignored by most current tools due to reasons of computational complexity and scarcity of these motifs.

We do not consider pseudoknots here. However, several comparative approaches that include pseudoknots

are currently under development, and certainly merit a comparative study of their own. Note that in an

application scenario, we often do not know whether the considerations (1–3) apply.

The comparative approach to structure inference is initiated from a set of homologous RNA sequences.

Attempts are made to infer the in-vivo structure for each of them, as well as a consensus structure that

captures the common, relevant structural aspects. The consensus structure per se does not exist in vivo,

and so some mathematical rigour should be applied when working with this notion.

An RNA sequence is a string over the RNA alphabet {A,C,G,U}. An RNA sequence B = b1, . . . , bn

contains n bases, but no structural information. For comparative analysis, we are given the RNA sequences

B1, . . . , Bk. A secondary structure can be associated with each sequence B as a string S over the alphabet

{”(”, ”.”, ”)”}, where parentheses in S must be properly nested, and B and S must be compatible: If

(si, sj) = (”(”, ”)”), then (bi, bj) must be a legal base pair. A base pair is also denoted as bi · bj , si · sj , or

simply i · j when the sequence is clear from the context. Both sequences and structures may be padded

with a gap symbol ”-”, in order to align sequences and structures of different lengths. For compatibility of

padded sequences and structures, we require that bi = ”-” iff si = ”-”.

A multiple structural alignment is a multiple sequence alignment of the 2 ∗ k sequences, B1, S1, . . . , Bk, Sk,

such that Bi is compatible with Si, and the following consistency criterion is satisfied: For any S i and Sj

and any base pair sik · sil, we have sjk 6= ”)” and sjl 6= ”(”, and if sjk = ”(” or sjl = ”)”, then sjk · sjl . This

means that if one partner of a base pair in Sj is aligned to one partner in Si, their partners must also be

aligned to each other. A violation of consistency is shown below (left), together with a possible correction

(right).

AACCAAAAAGAGAA AACCA-AAAAGAGAA

..((.....).).. ..((.-....).)..

AACUUAAAAGAGAA AA-CUUAAAAGAGAA

..(.(...)).... ..-(.(...))....

Note that the inconsistent alignment may maximise sequence similarity, showing 3 mismatches versus 1

mismatch and 2 indels, with the concrete outcome depending on the gap scoring used. Inconsistency is the

4

Page 5: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

reason why it is dangerous to align two structures in string representation by a standard sequence

alignment algorithm. Inconsistency is hard to detect by human eye inspection, and structural alignments

in data bases are not always free from consistency violations.

A consensus structure C for a multiple structural alignment can be determined by a majority rule

approach using a threshold p with 0.5 < p ≤ 1. We define ck = x if sik = x for at least dp ∗ ke sequences Si,

and ck = ”.”, otherwise. The latter definition is somewhat arbitrary; when relating the consensus structure

to a particular sequence B in the alignment, we quietly turn those dots into gaps that align with gaps in B.

For p = 1, we speak of a strict consensus, and the base pair set in C is the intersection of the base pairs in

all Si.

A consensus structure exhibits base pairs shared by the majority of structures under consideration, but has

no sequence information associated with it. Each individual structure for a concrete sequence typically has

additional base pairs which are properly nested between those that constitute the consensus. Given a

consensus structure C and a sequence B compatible with it, we can obtain a structure refold(B,C) which

is the best thermodynamic folding for B that exhibits the base pairs specified by C, plus additional ones

that do not conflict with the former. Refolding can be achieved by RNAfold with option −C (if B and S

contain gaps, we remove them before refolding and reintroduce them in the same positions afterwards).

Given a consistent structural alignment, it is easy to derive a consensus structure, as we can count

majorities at individual positions. If the 5’ partner of a base pair passes the majority threshold,

consistency implies that its 3’ partner also makes it into the consensus.

Given a consensus structure and a sequence alignment without structural information, we can approximate

a structural alignment by computing Si = refold(Bi, C). We call this structural alignment reconstruction.

While all Si will be consistent with C, and with each other as far as the base pairs of C are concerned,

they may be inconsistent for the base pairs introduced in refolding. This is tolerable, since if we trust the

consensus to capture the relevant common structural features, there is no need to require that all members

of a family agree upon extra-consensus features.

We note in passing that it seems worthwhile to study the conditions under which consensus derivation and

structural alignment reconstruction are mutually inverse operations, but such theoretical issues are outside

our present scope.

5

Page 6: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Interpreting data base information

While the plans A, B and C we are about to evaluate strive to find a good consensus structure from

sequence data, the “truth” available to us comes in a different form. Structural data bases only convey a

consensus by example: They provide a reference sequence, say B1, with an experimentally proved structure

S1, and provide a multiple sequence alignment of B1, S1 and additional sequences B2, . . . , Bn in the family

under consideration. The sequence alignment is chosen to exhibit structural similarities between the

reference structure and the other family members, but in general, we do not know the precise model of

achieving similarity, nor do we know whether this model has been solved to optimality.

One consequence of this situation would be to conclude that the reference structure is the only reliable

anchor point available to us for evaluation. Comparative analysis tools would then be evaluated by their

capacity to predict this particular structure by using family information. This would be a meaningful way

to proceed, however, the effect of structural homogeneity within a sequence family would go unmeasured,

and so would the difficulty or success of exploiting it. We therefore proceed in a different way which we call

consensus reconstruction.

The reference structure S1 need not be compatible with any Bi except for i = 1. However, we can still

compute Si := refold(Bi, S1) by treating bases as unpaired where they violate compatibility. (This is also

achieved with RNAfold, option -C.) What we obtain in this way is a reconstructed structural alignment,

which will be consistent to the extent that the reference structure indeed describes the common structural

features, and to the extent that the database sequence alignment reflects these. In all our test cases, this

alignment was overall consistent, an indicator that the families and their structural features are in fact well

defined. From this alignment, we derive a consensus structure as explained above using a threshold p = 0.5,

which will serve as the standard of truth in our evaluation.

One may argue that our approach to reconstruct the truth is somewhat ad-hoc and should be replaced by

a more systematic method. However, this is what the tools we evaluate try to achieve, and we should not

add one of our own as the standard of truth. Hence, our consensus reconstruction is designed to stay as

close as possible to the data base information.

Test data-sets

We have compiled RNA sequence alignments consisting of up to 11 sequences derived from reliable sources

(see table 1). These are subsequently used to test several RNA analysis packages. Each alignment contains

at least one reference sequence B1 with (preferably) an experimentally verified secondary structure S1.

6

Page 7: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Experimental verification of a structure may be from a variety of sources: x-ray crystallography, NMR,

enzymatic structure probing or phylogenetic inference. A comparison of phylogenetic with x-ray

crystallographic structures has shown the phylogenetic predictions of rRNA to be very reliable (sensitivity

> 97%) [61]. This data specifies a “consensus by example”, as explained above, to which our consensus

reconstruction was applied to obtain the “true” consensus.

To avoid results bias, we constructed test alignments, with corresponding phylogenies that, wherever

possible, were free of highly similar clades. In addition, we endeavoured to ensure that the reference

sequence was central to the phylogeny, or more specifically, not an outgroup. To meet these requirements,

sequences from large data-sets were sorted into high-similarity and medium-similarity groups (with respect

to the model sequence), from which maximum-likelihood phylogenies [62] were constructed. These were

pruned until the desired size and topology was achieved. For each data-set two sequence alignments were

constructed, one of high sequence identity (approximately 90-99%) and the other more diverse data-set of

medium sequence identity (approximately 70-90%). For pursuing Plan C, we chose a scoring function that

ignores sequence information in the structure alignment step – this allowed us to evaluate the case of

lacking sequence conservation using the same data-sets.

Our data-sets are quite diverse and must for the purposes of this study be considered difficult to analyse in

structural terms. The shape of ribosomal RNA is believed to be influenced by interaction with ribosomal

proteins. The shape of RNAse P shows relatively little sequence and structure conservation, and

furthermore, it contains pseudoknots which are generally excluded by prediction algorithms. Transfer

RNAs are known to be a hard case for thermodynamic folding, primarily due to the propensity of modified

bases which influence structure formation. All tools tested may perform better upon less complex

data-sets, but the purpose of this study is not to show how good the algorithms are but to compare

relative performance when things are not easy.

Performance Measures

Sensitivity (X) and selectivity (Y ) are common measures for determining the accuracy of prediction

methods [63]. Selectivity is also known as “specificity” or “positive predictive value” [64]. We use slightly

modify versions of the standard definitions of X and Y for examining RNA secondary structure prediction:

X :=TP

TP + FNY :=

TP

TP + (FP − ξ)

where TP is the number of “true positives” (correctly predicted base-pairs), FN is the number of “false

7

Page 8: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

negatives” (base-pairs in the reference structure that were not predicted) and FP is the number of “false

positives” (in-correctly predicted base-pairs). However, not all FP base-pairs are equally false! We classify

FP base-pairs as either inconsistent, contradicting or compatible. Predicted base-pairs which conflict with

a base-pair in the reference structure are labelled inconsistent (i.e. i · j is predicted where either i · k

and/or h · j are paired in the reference structure (h 6= i and j 6= k)). Predicted base-pairs (i · j) which are

non-nested with respect to the reference structure are labelled contradicting (i.e. there exists base-pairs

k · l in the reference satisfying k < i < l < j). Note that some base-pairs may both contradict and be

inconsistent with the reference structure. Predicted base-pairs which are neither true positive, contradicting

or inconsistent are labelled compatible and can be considered neutral with respect to algorithm accuracy.

Hence these are subtracted in the selectivity evaluation, their number is ξ in the above equation.

It is of interest to note that the base-pair metric [65, 66] between the reference and predicted structures

dBP (Sref , Spred) is the sum of FN and FP , and hence is different from the measure used here.

A measure combining both selectivity and sensitivity is useful for ranking algorithms. For this we employ

the Matthews correlation coefficient [63] defined below:

MCC =TP × TN − (FP − ξ)× FN√

(TP + FP − ξ)(TP + FN)(TN + FP − ξ)(TN + FN)

MCC ranges from −1 for extremely inaccurate (TP = TN = 0) to 1 for very accurate predictions

(FP − ξ = FN = 0). When comparing RNA structures TN = 0 occurs only in extreme examples, hence

MCC generally ranges from 0 to 1. Furthermore, MCC can be approximated by the arithmetic-mean or

geometric-mean of X and Y .

Caveats

Results of observations based on the above measures must be interpreted with care. We list a number of

caveats that must be kept in mind when proceeding to the subsequent sections.

Use of defaults: In all tests, one could possibly obtain better predictions by tuning the program’s

parameters. We felt that it would be inappropriate to do so, since in the evaluation, we know the correct

result and could use this knowledge in the tuning, whereas in a true application context, one does not have

such guidance. Hence we used the recommended defaults in all cases.

Tool abuse: In some cases we apply a tool to data where we know that the model structure has features

not recognised by the tool. An example is a structure with multiloops or pseudoknots, searched for with a

tool that explicitly excludes such structures. We permit such cases, because again, in a true application

8

Page 9: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

context one does not know whether the tool is appropriate or not, and it is still of interest to see how close

to the correct structure one can get.

Standard of truth: We take for granted the correctness of structural alignments taken from the literature,

and the consensus reconstructed thereof. Should one of the tested algorithms produce a result that is

actually better (closer to the functionally important structure), it will be penalised. Also, we do not

consider a large number of data-sets here, it is possible that performance of some algorithms improves on a

different selection of data-sets.

Tools improve: Our data reflect the state of the art in 2004. Most of the tools tested are very recent, and

their authors are still improving them. Hence, not all observations will remain reproducible. In fact, we

hope this study helps to obtain better results in the future.

ResultsSingle Sequence Methods

The accuracy of the MFE single sequence method has been evaluated elsewhere and was found to have an

accuracy of 73% when averaged over many different RNAs and “base-pair slippage” was tolerated in the

evaluation [67]. A recent and more stringent work found MFE predictions had a sensitivity of 56% and

selectivity of 46% for RNase P, SRP and tmRNA structures [64]. Similar values are also reported by the

“Gutell Lab” for tRNA and rRNA structures [68–70]. We need to clarify the accuracy of this method on

the particular data-sets we employ here for comparison with the multi-sequence methods. After all, if MFE

folding worked perfectly for our given data-sets, there would be no need to resort to comparative methods.

Mfold & RNAfold

Mfold [12] and RNAfold [13] both implement the Zuker-Stiegler algorithm for computing minimal free

energy (MFE) structures assuming a “nearest neighbour model” and using empirical estimates of

thermodynamic parameters for neighbouring interactions and loop entropies to score structures. The

algorithm is O(n3) in time and O(n2) in memory where n is the sequence length. Both employ the same

thermodynamic parameters [67]. Hence, differences in the predictions are generally minor and are the

result of slightly different implementations. There appears to be no significant differences in terms of

algorithm accuracy.

The sensitivity, selectivity and correlation of MFE methods (for the four data-sets considered here) ranged

from 22-63%, 20-60% and 0.18-0.61 respectively (See figures ?? & ??). The low accuracies (22%, 20% &

9

Page 10: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

0.18) are due to an alternative long-stem conformation of S.cerevisiae tRNA-PHE which the free energy

methods favour. Mfold infers ‘suboptimal’ structures by calculating minimum free energy structures with

the restriction that every possible base-pair is forced in a one-by-one fashion. Unique structures are then

ranked by energy. Investigating the top two suboptimal structures from Mfold resulted in increases in the

ranges of sensitivity, selectivity and correlation, 22-69%, 20-67% and 0.18-0.68 respectively. The

predictions shown here are used to illustrate the potential advantages of using comparative analyses over

single sequence methods.

Sfold

Sfold [41] represents another energy-based single-sequence folding algorithm. For a given RNA, Sfold

stochastically samples all possible structures in the Boltzmann ensemble of secondary structures using

conditional probabilities which are computed with the partition function [71]. Clustering techniques could

then be used to obtain representative ‘likely’ structures. Instead, the current implementation samples 1000

structures, sorts these by energy, the minimum and maximum energy structures are computed and the

energy range divided into 10 equally sized energy blocks. The minimum energy structure from each block

is returned with ranking 1 to 10. We consider the top 3 structures labelled ‘Sfold (1-3)’. In terms of

accuracy, the results are very similar to those of the Zuker-Stiegler single sequence methods, although with

a slightly higher variance (See figures ?? & ??).

Intrinsic limits of single sequence methods

There are systematic limits to the accuracy of single sequence prediction methods. The thermodynamics

may not be accurate, as some parameters are extrapolated and parameter measuring conditions in vitro are

different from in vivo conditions. Indeed the thermodynamic model itself is an estimate of the real physics

of RNA folding. Also, many bases of structural RNAs are chemically modified by sugar methylation,

pseudo-uridine, dihydrouracil, etc, these are generally ignored by these methods. There has been some

recent progress here however [72]. The kinetics of folding are ignored. Given only a single sequence, we

have no way to distinguish base pairs and structure elements important for the consensus from those that

are peculiar for the given sequence. Finally, some functional RNAs have bistable structures, while in

others, the structure is irrelevant, hence not conserved, and the optimal MFE structure is biologically

meaningless. These are some of the reasons why researchers proceed to comparative methods.

10

Page 11: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Comparative Method: alignment folding (Plan A)RNAalifold

RNAalifold [21] implements an extension of the Zuker-Stiegler algorithm for computing a consensus

structure from RNA alignments. The algorithm computes an averaged energy matrix E ij =N∑k

Ekij (where

N is the number of sequences in the alignment) and a covariation score matrix, augmented with penalties

for inconsistent sequences, Bij . A standard trace-back procedure is performed to recover a consensus

structure with the optimal sum-of-average-energy-and-covariation-score E ij +Bij . The algorithm is

remarkably efficient O(N · n2 + n3) in time and O(n2) in memory.

The sensitivity, selectivity and correlation of the RNAalifold predictions ranged from 57-91%, 57-100% and

0.57-0.95 respectively, showing a significant increase in the accuracy measures when compared to the

MFE-methods.

Pfold

Pfold implements a “stochastic context free grammar” (SCFG) designed to produce a “prior probability

distribution of RNA structures” for a RNA alignment input [23,24]. A maximum-likelihood phylogeny is

used to weight posterior probabilities computed from large reference data-sets.

The algorithm is generally very accurate and efficient. Hence, the over-all sensitivity, selectivity and

correlation of the Pfold predictions ranged from 0-100%, 0-100% and 0.0-1.0, respectively. But removing

those points where Pfold predictions were empty structures (LSU rRNA (H & M) and SSU rRNA (M), see

figure ??), the prediction accuracies ranged from 66-100%, 89-100% and 0.77-1.0, respectively. The zeros

are due to ‘under-flow errors’, a solution is presently under construction by the authors (pers. commun.

Bjarne Knudsen).

ILM

ILM (iterated loop matching) is one of the few comparative RNA folding algorithms which can return

pseudo-knotted structures [22]. It uses a combination of thermodynamic and mutual information content

scores [18] to produce a secondary structure. All possible stems (allowing “small” internal loops and

bulges) are generated and ranked according to a combination of thermodynamic and mutual-information

scores. The stem with maximal score is selected, scores are updated and stems conflicting with the selection

removed, then the next highest scoring stem is selected. This algorithm is iterated until no stems remain.

ILM generally ranked low in terms of selectivity and was not as sensitive as either RNAalifold or Pfold on

11

Page 12: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

the high similarity data, but did improve on the medium similarity data-sets (see figure ??). The over-all

sensitivity, selectivity and correlation of ILM predictions ranged from 44-100%, 37-75% and 0.40-0.86,

respectively. To ensure the low selectivity values weren’t due to the representative-reference-structure

being pseudo-knot free we re-evaluated ILM with reference structures replete with pseudo-knots. The new

sensitivity, selectivity and correlation values ranged from 31-100%, 26-75% and 0.29-0.86, in fact evaluating

with pseudo-knotted structures did little to increase ILM selectivity.

Comparative Method: simultaneous sequence alignment and folding (Plan B)Sankoff

The Sankoff algorithm is a dynamic programming approach to obtain a common base-pair list with

maximal sum of base-pair weights. Basically, this is a merger of sequence alignment and Nussinov [73]

(maximal-pairing) folding dynamic programming methods [26]. Sankoff’s algorithm can be used to obtain

both an alignment and consensus structure. Full implementations of the “Sankoff algorithm” for the

solution of simultaneous RNA folding, alignment and protosequence problems have proven too

computationally taxing (O(n6) in time, and O(n4) in space) to be practical [25]. Hence, three restricted

versions of this algorithm have been implemented. These are Foldalign [27], Dynalign [29] and recently

PMmatch has also been published [26]. Carnac [74] is another recent innovation designed to detect

conserved stems in unaligned sequences, we include it here as a relative of the Sankoff approach.

Foldalign

Foldalign [27] can be interpreted as “a mixture of local alignment and maximum number of base-pairs

algorithm” [28]. A combination of “clustal” [15] and “consensus” [75] heuristics are used to build multiple

sequence alignments from pair-wise comparisons. Restricting maximum motif size (for this study 50 was

used) and forbidding branched structures (multi-loops) reduces the time complexity to O(n4N) in time

(where N is the number of sequences and n is the length of the longest sequence). A simple match-based

scoring scheme is used to rank putative conserved structure elements.

The Tool Abuse Caveat generally applies to the tool Foldalign as all of our data-sets contain multi-loops.

The use of Foldalign for the prediction of global, multi-looped secondary structures is not recommended. It

is designed for the location of short regulatory motifs such as IREs [76] in messenger RNA. Hence the

rather poor sensitivity, selectivity and correlation, which ranged from 5-24%, 23-36% and 0.11-0.27

respectively, for our test datasets.

12

Page 13: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Dynalign

Dynalign is a pairwise implementation of the Sankoff algorithm, which uses a “full energy model” to locate

a common low energy structure (including multi-loops) and align two structural RNAs. The computational

complexity of the full Sankoff is reduced by restricting the “maximum insert size” to M . The complexity is

thus reduced to O(n4M3).

The current Dynalign implementation is restricted to pair-wise sequence comparisons. Rather than

compute all 12n(n− 1) pairwise foldings we compared all sequences with the reference structure. Due to the

computational expense of this algorithm it could only be used to predict tRNA and RNase P structures.

Dynalign performed well on the tRNA, medium sequence homology data-set (sensitivity, selectivity and

correlation of 94%, 95% and 0.94 respectively, when averaged over all pairwise alignments with the

reference sequence). With this one high-scoring point removed, averaged sensitivity, selectivity and

correlation values ranged from 32-54%, 33-54% and 0.32-0.54 respectively. Comparing the performances of

MFold and Dynalign showed that MFold performance was always superior on the RNase P data-set,

Dynalign however did much better on the shorter and more diverse tRNA sequences. Performance gains

could be made by investing more computer time and refolding RNase P with larger ‘maximum insert size’,

which was set to 10 during this study.

Carnac

The Carnac algorithm, as mentioned previously, is not strictly an implementation of the Sankoff algorithm.

A set of filters are employed through which sets of sequences are passed in a pair-wise fashion [74].

Sequences are scanned for stems and “high similarity” regions of sequences (dubbed “anchor points”) are

identified, a dynamic program is used to select conserved stems using anchor point and covariation

information.

The Carnac algorithm was remarkably selective at base-pair predictions. However, the sensitivity of the

algorithm was generally low, although when evaluated by the correlation coefficient Carnac was

comparable to RNAalifold and Pfold. Sensitivity, selectivity and correlation values for Carnac predictions

ranged from 45-71%, 92-100% and 0.65-0.82 respectively. The sensitivity of Carnac can be increased by

constraining a minimum free energy fold (i.e. with “RNAfold -C”) with the Carnac predicted structure.

On average this increased the sensitivity by 22.5, decreased the selectivity by 17.2 and slightly increased

the correlation by 0.05.

13

Page 14: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Alignment of predicted structures (Plan C)RNAforester

RNAforester [37] implements the tree alignment model. In contrast to approaches that produce only a

similarity value, but no underlying alignment, it computes pairwise alignments of two input structures.

RNAforester can produce either global or local alignments; we used the global mode. A structure

alignment is itself a branching (tree-like) structure; the set of matched base pairs can be derived from it

and evaluated as with the other approaches.

We used the tRNA and RNase P data-sets and generated structure single sequence predictions with

RNAfold. All predicted structures were aligned pairwise, and the highest scoring alignment was used to

derive a consensus that was evaluated against the consensus tRNA model structures.

Sensitivity, selectivity and correlation ranges of consensus structures computed from the highest scoring

RNAforester alignments were 29-67%, 27-67% and 0.26-0.66 respectively. It seems likely that much of the

inaccuracy of this approach is due to MFE structure prediction, however the structure-clustering approach

frequently separates mis-folded MFE predictions from the accurate folds.

MARNA

The MARNA algorithm [39] proceeds by constructing edge weights between nucleotides in a pairwise

fashion. Weights are structure-enhanced-sequence-similarities transformed from edit distances proposed by

Zhang [77]. Phase two pipes the set of alignment edges into t-coffee [16] for multiple alignment production.

The resultant alignments are not strictly structural alignments in the sense defined above. Rather, these

are sequence alignments influenced by structure.

Sensitivity, selectivity and correlation values of consensus structures computed from MARNA alignments

of MFE structures ranged from 29-52%, 32-84% and 0.30-0.65 respectively. We also trimmed high entropy

base-pairs from the MFE predictions using the bound Qij > 1, where Qij =√qiqj , qi =

∑j pij log pij , and

pij are pair-probabilities computed using McCaskill’s partition function [71]. The new accuracy ranges

were 29-71%, 92-100% and 0.53-0.84. A related approach for trimming of low probability was recently

shown to greatly improve the selectivity of MFE predictions [78]. MARNA is generally less dependant

upon the accuracy of the input structures hence performs slightly better with the poorly predicted tRNA

structures than RNAforester.

14

Page 15: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Discussion

We have evaluated three different strategies for comparative structure prediction, and altogether eight tools

(not counting the single sequence methods). The results of which are summarised in figures ?? & ??. A

surprising discovery given that the test data-sets are so diverse is that algorithm specific clusters formed in

sensitivity versus selectivity scatter plots, indicating algorithm-specific eccentricities. A number of

algorithms which might have been evaluated here have been excluded, primarily due to the heavy

computational costs of the various implementations on our longer data-sets. We favoured recent algorithms

which could be compiled on modern computers and those with input and output which could be simply

dealt with (for example returning dot-bracket [13,37,79] or tabular-connect type formats [12,29,41], rather

than coordinates and lengths of stacks or graphic (gif/pdf) representations favoured by a minority of

researchers).

Practical recommendations

For well aligned short sequences, both Pfold and RNAalifold generally perform well, PFold performed

marginally better than RNAalifold. It is likely that some moderate refinements to RNAalifold would

improve accuracy without altering the efficiency, for example, if gaps were not penalised in the free-energy

evaluation and a more sophisticated model for scoring mutations was employed, perhaps ribosum

matrices [80] could be used to weight base-pair bonuses and penalties. For well aligned, long sequences the

performance and speed of RNAalifold was excellent. For data-sets consisting of short (< 200 bases) and

diverse sequences Dynalign might do well or one might choose to use a mixture of RNAalifold or Pfold to

fold similar clades and RNAforester or MARNA to align folded clades.

Advocates of plan A should note that many multiple sequence alignment algorithms generally do not

favour transitions over transversions or employ ad hoc 2-parameter methods to model these (clustalw [15]

for example). Structural RNA sequences however evolve rapidly via structure neutral mutations which are

frequently transitions and rarely transversions [80,81]. Multiple sequence algorithms which employ more

complex yet more accurate models of sequence evolution will undoubtedly produce “better” alignments for

folding.

Carnac produced highly selective structures for all the test data-sets, which if used to constrain a free

energy fold produced sensitive predictions with a small cost in selectivity. The consistency of Carnac

performance is remarkable, for all the data-sets considered here this heuristic approach performed well. It

is however unclear how Carnac performs on highly diverse data-sets.

15

Page 16: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

For advocates of plan C, we have an encouraging message: Both MARNA and RNAforester perform better

on the medium similarity data than on high similarity data. This seems paradoxical at first glance, but one

must understand that for an approach purely based on predicted structures, high sequence similarity can

be a curse rather than a blessing: If sequences are very similar, they may jointly fold into the wrong MFE

structure. With more sequence variation, it becomes more likely that at least some family members have

good predictions, which by their mutual similarity can be picked out from the rest. This means that

especially in the case of low sequence similarity, where nothing else works, plan C, currently the least

explored strategy of all, has a certain promise.

Future Directions

An implementation of the single sequence pseudoknot algorithms [42,43,82] employing similar strategies to

RNAalifold [21] for alignment folding would be useful. Based upon the RNAalifold results this approach

would dramatically increase the accuracy of these algorithms upon certain data-sets. Also, an extension of

these allowing constrained foldings to incorporate prior knowledge would be of assistance, this has proved

extremely useful for MFE predictions. Sampling structures from reference alignments is also likely to prove

beneficial.

The implementation of fast and accurate variants of the Sankoff algorithm remains an open problem.

Again allowing constrained foldings and alignments would be useful. The further development of

“BLAST-like” folding heuristics for this should be a priority, obviously Carnac is a good start.

The MARNA approach for producing structurally enhanced multiple alignments produced rather selective

results after trimming high-entropy base-pairs from MFE predictions. This suggests that weighting

edit-distances with partition-function derived probabilities or entropies will produce reasonable RNA

alignments. A consensus structure could then be derived from MFE-structures or from PFold or

RNAalifold predictions. This approach would effectively decouple the Sankoff algorithm into manageable

structure-enhanced-alignment and folding stages.

More training data is essential for this field to progress, for this homology search tools are essential.

Infernal [79] used to construct the Rfam database [83] is an excellent beginning but sensitivity might be

increased with a phylogenetic approach and RNA-specific sequence search tools.

16

Page 17: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Authors’ contributions

PPG carried out the experiments, the analysis and drafted the manuscript. RG suggested comparing

comparative structure prediction methods and assisted in the manuscript preparation. All authors read

and approved the final manuscript.

Acknowledgements

The authors thank the numerous researchers who provided access, documentation and installation

assistance for their algorithms; Notably Ivo Hofacker, Dave Mathews, Bjarne Knudsen, Matthias

Hochsmann and Sven Siebert, authors of RNAalifold, Dynalign, Pfold, RNAforester and MARNA

respectively. PPG thanks Niels Hansen and Andreas Wilm for useful discussions and advice. PPG was

supported by a DFG (German Research Foundation) post-doctoral scholarship and a Carlsberg Foundation

Grant (21-00-0680). The basis of much of this work was conceived at the ESF and NIH funded 2003

computational RNA workshop in Benasque, Spain.

References1. Doudna J, Cech T: The Natural chemical repertoire of natural ribozymes. Nature 2002, 418:222–228.

2. Poole AM, Jeffares DC, Penny D: The path from the RNA world. Journal of Molecular Evolution 1998,46:1–17.

3. Jeffares DC, Poole AM, Penny D: Relics from the RNA world. Journal of Molecular Evolution 1998,46:18–36.

4. Kapranov P, Cawley SE, Drenkow J, Bekiranov S, Strausberg RL, Fodor SPA, Gingeras TR: Large-scaletranscriptional activity in chromosomes 21 and 22. Science 2002, 296:916–919.

5. Kampa D, Cheng J, Kapranov P, Yamanaka M, Brubaker S, Cawley S, Drenkow J, Piccolboni A, Bekiranov S,Helt G, Tammana H, Gingeras TR: Novel RNAs Identified From an in-depth analysis of thetranscriptome of human chromosom es 21 and 22. Genome Research 2004, 14:331–342.

6. Cawley S, Bekiranov S, Ng H, Kapranov P, Sekinger E, Kampa D, Piccolboni A, Sementchenko V, Cheng J,Williams A, Wheeler R, Wong B, Drenkow J, Yamanaka M, Patel S, Brubaker S, Tammana H, Helt G, StruhlK, Gingeras T: Unbiased Mapping of Transcription Factor Binding Sites along HumanChromosomes 21 and 22 Points to Widespread Regulation of Noncoding RNAs. Cell 2004,116(4):499–509.

7. Mattick J, Gagen M: The Evolution of Controlled Multitasked Gene Networks: The Role of Intronsand Other Noncoding RNAs in the Development of Complex Organisms. Mol. Biol. Evol. 2001,18:1611–1630.

8. Mattick J: Non-coding RNAs: the architects of eukaryotic complexity. EMBO Reports 2001,2:986–991.

9. Tinoco I, Bustamante C: How RNA folds. J Mol Biol 1999, 293(2):271–281.

10. Onoa B, Tinoco I: RNA folding and unfolding. Curr Opin Struct Biol 2004, 14(3):374–379.

11. Furtig B, Richter C, Wohnert J, Schwalbe H: NMR spectroscopy of RNA. Chembiochem 2003,4(10):936–962.

12. Zuker M, Stiegler P: Optimal computer folding of large RNA sequences using thermodynamics andauxiliary information. Nucleic Acids Research 1981, 9:133–148.

17

Page 18: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

13. Hofacker IL, Fontana W, Bonhoeffer S, Stadler PF: Fast Folding and Comparison of RNA SecondaryStructures. Monatshefte fur Chemie 1994, 125:167–188.

14. Woese C, Pace N: The RNA World, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY 1993 chap.Probing RNA structure, function, and history by comparative analysis, :91–117.

15. Thompson J, Higgins D, Gibson T: CLUSTAL W: improving the sensitivity of progressive multiplesequence alignment through sequence weighting, positions-specific gap penalties and weightmatrix choice. Nucleic Acids Research 1994, 22:4673–4680.

16. Notredame C, Higgins D, Heringa J: T-COFFEE: A novel method for fast and accurate multiplealignment. JMB 2000, 302:205–217.

17. Gotoh O: Multiple sequence alignment: algorithms and applications. Adv. Biophys. 1999, 36:159–206.

18. Chiu DK, Kolodziejczak T: Inferring consensus structure from nucleic acid sequences. Comput. Appl.Biosci. 1991, 7:347–352.

19. Gutell RR, Power A, Hertz GZ, Putz EJ, Stormo GD: Identifying constraints on the higher-orderstructure of RNA: continued development and application of comparative sequence analysismethods. Nucleic. Acids. Res. 1992, 20:5785–5795.

20. Gorodkin J, Heyer L, Brunak S, Stormo G: Displaying the information contents of structural RNAalignments. Bioinformatics 1997, 13:583–586.

21. Hofacker I, Fekete M, Stadler P: Secondary Structure Prediction for Aligned RNA Sequences. Journalof Molecular Biology 2002, 319(5):1059–1066.

22. Ruan J, Stormo G, Zhang W: An Iterated loop matching approach to the prediction of RNAsecondary structures with pseudoknots. Bioinformatics 2004, 20:58–66.

23. Knudsen B, Hein J: Pfold: RNA secondary structure prediction using stochastic context-freegrammars. Nucleic Acids Research 2003, 31(13):3423–3428.

24. Knudsen B, Hein J: RNA secondary structure prediction using stochastic context-free grammarsand evolutionary history. Bioinformatics 1999, 15(6):446–454.

25. Sankoff D: Simultaneous solution of the RNA folding, alignment and protosequence problems.SIAM Journal on Applied Mathematics 1985, 45:810–825.

26. Hofacker I, Bernhart S, Stadler P: Alignment of RNA Base Pairing Probability Matrices.Bioinformatics 2004.

27. Gorodkin J, Heyer L, Stormo G: Finding the most significant common sequence and structure motifsin a set of RNA sequences. Nucleic Acids Research 1997, 25(18):3724–3732.

28. Gorodkin J, Stricklin SL, Stormo GD: Discovering common stem-loop motifs in unaligned RNAsequences. Nucleic Acids Research 2001, 29(10):2135–2144.

29. Mathews D, Turner D: Dynalign: An Algorithm for Finding the Secondary Structure Common toTwo RNA Sequences. Journal of Molecular Biology 2002, 317(2):191–203.

30. Tai K: The tree-to-tree correction problem. Journal of the ACM 1979, 26:422–433.

31. Shapiro BA: An algorithm for comparing multiple RNA secondary structures. CABIOS 1988,4:381–393.

32. Shapiro B, Zhang K: Comparing multiple RNA secondary structures using tree comparisons.CABIOS 1990, 6:309–318.

33. Zhang K, Shasha D: Simple fast algorithms for the editing distance between trees and relatedproblems. SIAM Journal of Computing 1989, 18(6):1245–1262.

34. Wang L, Jiang T, Gusfield D: A More Efficient Approximation Scheme for Tree Alignment. SIAM J.Comput. 2000, 30:283–299.

35. Jiang T, Wang J, Zhang K: Alignment of Trees - An Alternative to Tree Edit. Theor. Comput. Sci.1995, 143:137–148.

36. Sczyrba A, Kruger J, Mersch H, Kurtz S, Giegerich R: RNA-related tools on the BielefeldBioinformatics Server. Nucleic Acids Research 2003, 31(13):3767–3770.

18

Page 19: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

37. Hochsmann M, Toller, Giegerich R, Kurtz S: Local Similarity of RNA Secondary Structures. Proc. ofthe IEEE Bioinformatics Conference 2003, :159–168.

38. Wang Z, Zhang K: Alignment between Two RNA Structures. Lecture Notes in Computer Science 2001,2136:690–703.

39. Siebert S, Backofen R: MARNA: A Server for Multiple Alignment of RNAs. In In Proceedings of theGerman Conference on Bioinformatics 2003:135–140.

40. Ji Y, Xu X, Stormo G: A graph theoretical approach to predict common RNA secondary structuremotifs including pseudoknots in unaligned sequences. Bioinformatics Advance Access 2004, 20.

41. Ding Y, Lawrence C: A statistical sampling algorithm for RNA secondary structure prediction.Nucleic Acids Research 2003, 31(24):7280–7301.

42. Dirks R, Pierce N: A Partition Function Algorithm for Nucleic Acid Secondary Structure,including Pseudoknots. Journal of Computational Chemistry 2003, 24:1664–1677.

43. Reeder J, Giegerich R: Design, implementation and evaluation of a practical pseudoknot foldingalgorithm based on thermodynamics. BMC Bioinformatics 2004, 5:104–104.

44. Venclovas C, Zemla A, Fidelis K, Moult J: Some measures of comparative performance in the threeCASPs. Proteins 1999, Suppl 3:231–237.

45. Venclovas C, Zemla A, Fidelis K, Moult J: Comparison of performance in successive CASPexperiments. Proteins 2001, Suppl 5:163–170.

46. Fischer D, Rychlewski L, Dunbrack R, Ortiz A, Elofsson A: CAFASP3: the third critical assessment offully automated structure prediction methods. Proteins 2003, 53(6):503–516.

47. Venclovas C, Zemla A, Fidelis K, Moult J: Assessment of progress over the CASP experiments.Proteins 2003, 53(6):585–595.

48. Burset M, Guigo R: Evaluation of gene structure prediction programs. Genomics 1996, 34(3):353–367.

49. Pavy N, Rombauts S, Dehais P, Mathe C, Ramana DV, Leroy P, Rouze P: Evaluation of gene predictionsoftware using a genomic data set: application to Arabidopsis thaliana sequences. Bioinformatics1999, 15(11):887–899.

50. Guigo R, Agarwal P, Abril JF, Burset M, Fickett JW: An assessment of gene prediction accuracy inlarge DNA sequences. Genome Res 2000, 10(10):1631–1642.

51. Thompson J, Plewniak F, Poch O: A comprehensive comparison of multiple sequence alignmentprograms. Nucleic Acids Res 1999, 27(13).

52. Thompson J, Plewniak F, Poch O: BAliBASE: a benchmark alignment database for the evaluation ofmultiple alignment programs. Bioinformatics 1999, 15:87–88.

53. Bahr A, Thompson J, Thierry J, Poch O: BAliBASE (Benchmark Alignment dataBASE):enhancements for repeats, transmembrane sequences and circular permutations. Nucleic Acids Res2001, 29:323–326, [{http://www.pmbrowser.info/pmdisplay.cgi?uids=11125126}].

54. Lassmann T, Sonnhammer E: Quality assessment of multiple alignment programs. FEBS Lett 2002,529:126–130, [{http://www.pmbrowser.info/pmdisplay.cgi?uids=12354624}].

55. Mandal M, Breaker R: Gene regulation by riboswitches. Nat Rev Mol Cell Biol 2004, 5(6):451–463.

56. Soukup J, Soukup G: Riboswitches exert genetic control through metabolite-inducedconformational change. Curr Opin Struct Biol 2004, 14(3):344–349.

57. Vitreschak A, Rodionov D, Mironov A, Gelfand M: Riboswitches: the oldest mechanism for theregulation of gene expression? Trends Genet 2004, 20:44–50.

58. Giegerich R, Haase D, Rehmsmeier M: Prediction and visualization of structural switches in RNA.Pacific Symposium on Biocomputing 1999, 4:126–137.

59. VoßB, Meyer C, Giegerich R: Evaluating the predictability of conformational switching in RNA.Bioinformatics 2004, 20(4).

60. Bengert P, Dandekar T: Riboswitch finder–a tool for identification of riboswitch RNAs. Nucleic AcidsRes 2004, 32(Web Server issue):154–159.

19

Page 20: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

61. Gutell R, Lee J, Connone J: The accuracy of ribosomal RNA comparative structure models. Curr.Opin. Struct. Biol. 2002, 12:301–310.

62. Felsenstein J: PHYLIP (Phylogeny Inference Package) version 3.6a3. Department of Genome Sciences,University of Washington, Seattle 2002. [Distributed by the author].

63. Baldi P, Brunak S, Chauvin Y, Andersen C, Nielsen H: Assessing the accuracy of prediction algorithmsfor classication: an overview. Bioinformatics 2000, 16:412–424.

64. Dowell R, Eddy S: Evaluation of several lightweight stochastic context-free grammars for RNAsecondary structure prediction. BMC Bioinformatics 2004, 5:71–71,[{http://www.pmbrowser.info/pmdisplay.cgi?uids=15180907}].

65. Fontana W, Konings D, Stadler P, Schuster P: Statistics of RNA secondary structures. Biopolymers 1993,33(9):1389–1404.

66. Moulton V, Zuker M, Steel M, Pointon R, Penny D: Metrics on RNA secondary structures. Journal ofComputational Biology 2000, 7(1-2):277–292.

67. Mathews D, Sabina J, Zuker M, Turner H: Expanded sequence dependence of thermodynamicparameters provides robust prediction of RNA secondary structure. J. Mol. Biol. 1999, 288:911–940.

68. Konings D, Gutell R: A comparison of thermodynamic foldings with comparatively derivedstructures of 16S and 16S-like rRNAs. RNA 1995, 1(6):559–574.

69. Fields D, Gutell R: An analysis of large rRNA sequences folded by a thermodynamic method. FoldDes 1996, 1(6):419–430.

70. Doshi K, Cannone J, Cobaugh C, Gutell R: Evaluation of the suitability of free-energy minimizationusing nearest-neighbor energy parameters for RNA secondary structure prediction. BMCBioinformatics 2004, 5:105–105.

71. McCaskill JS: The equilibrium partition function and base pair binding probabilities for RNAsecondary structures. Biopolymers 1990, 29:1105–1119.

72. Mathews D, Disney D, Childs M, Schroeder J, Zuker M, Turner D: Incorporating chemical modificationconstraints into a dynamic programming algorithm for prediction of RNA secondary structure.Proc. Natl. Acad. Sci. USA 2004, 101(19):7287–7292.

73. Nussinov R, Piecznik G, Grigg JR, Kleitman DJ: Algorithms for loop matchings. SIAM Journal onApplied Mathematics 1978, 35:68–82.

74. Perriquet O, Touzet H, Dauchet M: Finding the common structure shared by two homologous RNAs.Bioinformatics 2003, 19:108–116.

75. Hertz G, Hartzell G, Stormo G: Identification of consensus patterns in unaligned DNA sequencesknown to be functionally related. Comput. Appl. Biosci. 1990, 6:81–92.

76. Hentze M, Kuhn L: Molecular control of vertebrate iron metabolism: mRNA-based regulatorycircuits operated by iron, nitric oxide, and oxidativestress. Proc Natl Acad Sci U S A 1996,93:8175–8182.

77. Jiang T, Lin G, Ma B, Zhang K: A General Edit Distance between RNA Structures. Journal ofComputational Biology 2002.

78. Mathews D: Using an RNA secondary structure partition function to determine confidence in basepairs predicted by free energy minimization. RNA 2004, 10(8):1178–1190.

79. Eddy S: A memory efficient dynamic programming algorithm for optimal structural alignment ofa sequence to an RNA secondary structure. BMC Bioinformatics 2002, 3(18).

80. Klein R, Eddy S: RSEARCH: finding homologs of single structured RNA sequences. BMCBioinformatics 2003, 4:44–44.

81. Higgs PG: RNA secondary structure: physical and computational aspects. Quarterly Reviews ofBioPhysics 2000, 33(3):199–253.

82. Rivas E, Eddy S: The language of RNA: a formal grammar that includes pseudoknots.Bioinformatics 2000, 16(4):334–340.

20

Page 21: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

83. Griffiths-Jones S, Bateman A, Marshall M, Khanna A, Eddy SR: Rfam: an RNA family database. NucleicAcids Research 2003, 31:439–441.

84. Wuyts J, De Rijk P, Van de Peer Y, Winkelmans T, De Wachter R: The European Large SubunitRibosomal RNA database. Nucleic Acids Res 2001, 29:175–177.[http://oberon.fvms.ugent.be:8080/rRNA/lsu/].

85. Cannone J, Subramanian S, Schnare M, Collett J, D’Souza L, Du Y, Feng B, Lin N, Madabusi L, Muller K,Pande N, Shang Z, Yu N, Gutell R: The Comparative RNA Web (CRW) Site: An Online Database ofComparative Sequence and Structure Information for Ribosomal, Intron, and other RNAs.BioMed Central Bioinformatics 2002, 3(2). [http://www.rna.icmb.utexas.edu/].

86. Wuyts J, Van de Peer Y, Winkelmans T, De Wachter R: The European database on small subunitribosomal RNA. Nucleic Acids Res 2002, 30:183–185. [http://oberon.fvms.ugent.be:8080/rRNA/ssu/].

87. Brown J: The Ribonuclease P Database. Nucleic Acids Res 1999, 27:314–314.[http://www.mbio.ncsu.edu/RNaseP/home.html].

88. Kimura M: A simple model for estimating evolutionary rates of base substitutions throughcomparative studies of nucleotide sequences. Journal of Molecular Evolution 1980, 16:111–120.

21

Page 22: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

FiguresFigure 1 - RNA Analysis

Current automated approaches to analysing homologous RNA sequences and structures usually follow one

of three “plans”. Plan A uses aligned sequences (usually produced by a standard multiple sequence

alignment algorithm) to infer a consensus secondary structure from the evolutionary and energetic

information contained in an alignment. This is a highly successful approach, but is limited to data-sets

with sequence homology high enough for the alignment step to work yet divergent enough for detection of

structurally consistent mutations. Plan B employs the “Sankoff algorithm” to simultaneously align and

infer a consensus structure. This algorithm requires extreme amounts of memory and time. Plan C aligns

RNA structures rather than sequences. This approach can be used in the rare situation where reliable

structures are known. Representative algorithms which could be used for each plan are indicated within

the figure.

Figure 2 - Prediction correlation with reality

Matthews correlation coefficient versus the logarithm of the sequence length for a range of different

ncRNAs and structure prediction algorithms. Inset A shows accuracies of thermodynamic single sequence

prediction algorithms. Insets B and C shows accuracies of comparative methods on the high and medium

similarity data-sets respectively.

Figure 3 - ROC plots

We use ROC (receiver operating characteristic) plots to simultaneously display both sensitivity and

selectivity for plans A, B and C respectively. Accuracies of the MFE methods (MFold, RNAFold and

SFold) are shown in each plot to provide a base-line. Points on the line X = Y are as sensitive as they are

selective, points below this line indicates a greater selectivity, points above indicate greater sensitivity.

Points below the line X = 100− Y are worse than “random” assignments; Assuming base-pairs are

independent of each other (this is false for base-pairing). Points in the top right corner are “perfect”

predictions.

Interestingly many algorithms form characteristic clusters in these plots. Where the variance is sufficiently

small these have been indicated with a closed curve.

22

Page 23: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Data-set length mean pairwise Number of Alignment Structureseq. identity Sequences source source

High Med. High Med.

E.coli 2904 88.1 72.0 11 11 Wuyts et al., Cannone et al.,LSU rRNA (0.12) (0.35) (2001) (2002)

E.coli 1542 90.7 80.0 11 11 Wuyts et al., Cannone et al.,SSU rRNA (0.08) (0.21) (2002) (2002)

E.coli 377 81.5 67.1 9 11 Brown, (1999) Brown, (1999)RNase P (0.09) (0.41)

S.cerevisiae 73 84.4 60.0 11 11 Griffiths-Jones et al., SundaralinghamtRNA-PHE (0.19) (0.71) (2003) & Rao, (1975)

Table 1: Characteristics and sources of the four test data-sets, columns from left to right show: data-set,lengths, pairwise sequence similarity (mean pair-wise Kimura ”2-parameter” distance is shown in parentheses[88]), the number of sequences in each alignment and the alignment and structure sources are given.

23

Page 24: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Results Tables

The following tables display results of several structure predictions using a variety of algorithms upon

data-sets containing either S.cerevisiae tRNA-PHE, E.coli RNase P, E.coli SSU rRNA or E.coli

LSU rRNA sequences. Reading columns from left to right we show: prediction method, number of

base-pairs in the reference structure, number of base-pairs in the predicted structure, the number of true

positive base-pairs in the prediction (% sensitivity as described earlier in parentheses), the number of false

positive base-pairs in the prediction (% selectivity as described earlier in parentheses), correlation values

are the “Matthews correlation coefficient” (approximate correlation in parentheses).

24

Page 25: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Algorithm number of bps number of bps True Positives False Positives Correlationin reference in prediction (% sensitivity) (% selectivity) (%)

S.cerevisiae tRNA-PHE: Single Sequence Methods

RNAfold 18 23 4 (22.2) 16 (20.0) 0.178 (21.1)Mfold (1) 18 21 4 (22.2) 14 (22.2) 0.191 (22.2)Mfold (2) 18 22 8 (44.4) 11 (42.1) 0.409 (43.3)Mfold (3) 18 23 4 (22.2) 16 (20.0) 0.178 (21.1)Sfold (1) 18 23 4 (22.2) 16 (20.0) 0.178 (21.1)Sfold (2) 18 23 4 (22.2) 16 (20.0) 0.178 (21.1)Sfold (3) 18 21 4 (22.2) 14 (22.2) 0.191 (22.2)

Table 2:Each of these MFE-based attempts to predict the famous S.cerevisiae tRNA-PHE structure converges on an alternative lengthy-helix typestructure. Adding prior knowledge, such as forcing modified bases in the RNA sequence to be unpaired can produce dramatic improvements.

25

Page 26: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Algorithm number of bps number of bps True Positives False Positives Correlationin reference in prediction (% sensitivity) (% selectivity) (%)

S.cerevisiae tRNA-PHE: Comparative Methods

Plan A: ClustalW Alignment

RNAalifold (H) 21 20 19 (90.5) 0 (100.0) 0.950 (95.2)RNAalifold (H) + RNAfold -C 21 21 21 (100.0) 0 (100.0) 1.000 (100.0)RNAalifold (M) 18 14 14 (77.8) 0 (100.0) 0.880 (88.9)RNAalifold (M) + RNAfold -C 18 21 18 (100.0) 0 (100.0) 1.000 (100.0)ILM (H) 21 24 16 (76.2) 7 (69.6) 0.722 (72.9)ILM (M) 18 30 18 (100.0) 6 (75.0) 0.863 (87.5)Pfold (H) 21 21 20 (95.2) 0 (100.0) 0.975 (97.6)Pfold (M) 18 21 18 (100.0) 0 (100.0) 1.000 (100.0)

Plan B: Unaligned sequences

Carnac (H) 21 17 15 (71.4) 1 (93.8) 0.815 (82.6)Carnac (H) + RNAfold -C 21 21 19 (90.5) 1 (95.0) 0.925 (92.7)Carnac (M) 21 13 12 (57.1) 1 (92.3) 0.722 (74.7)Carnac (M) + RNAfold -C 21 22 16 (76.2) 5 (76.2) 0.757 (76.2)Dynalign (H) 21 22.40 11.50 (54.78) 10.20 (54.45) 0.5353 (54.59)Dynalign (M) 21 21.10 19.80 (94.27) 1.20 (95.00) 0.9448 (94.64)Foldalign (H) 21 16 5 (23.8) 11 (31.2) 0.259 (27.5)Foldalign (M) 21 16 5 (23.8) 10 (33.3) 0.268 (28.6)

Plan C: Structure alignment

MARNA (H) 21 19 6 (28.6) 12 (33.3) 0.295 (31.0)MARNA (M) 21 22 7 (33.3) 15 (31.8) 0.311 (32.6)MARNA-trim (H) 21 6 6 (28.6) 0 (100.0) 0.530 (64.3)MARNA-trim (M) 21 15 15 (71.4) 0 (100.0) 0.843 (85.7)RNAforester (H) 21 23 6 (28.6) 16 (27.3) 0.263 (27.9)RNAforester (M) 21 21 14 (66.7) 7 (66.7) 0.659 (66.7)

Table 3: Generally the comparative approaches perform much better than MFE methods at determining S.cerevisiae tRNA-PHE structure.For the consensus predictions of RNAalifold and Carnac we also computed “filled” structures using constrained MFE predictions. Thisusually improved the sensitivity of the methods. PFold uses a stem-extension procedure to fill structures.As the tRNA structure contains a multi-loop Foldalign is not expected to perform well here. Dynalign performed well on the most diversedata-set (M) but generally didn’t do well on the high similarity data-set.The structure alignment methods generally did poorly here. Most probably due to the miss-folded MFE structure which were used as input.Trimming the high entropy base-pairs from the input structures produced modest improvements.

26

Page 27: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Algorithm number of bps number of bps True Positives False Positives Correlationin reference in prediction (% sensitivity) (% selectivity) (%)

E.coli RNase P: Single Sequence Methods

RNAfold 110 116 69 (62.7) 46 (60.0) 0.612 (61.4)Mfold (1) 110 118 67 (60.9) 49 (57.8) 0.591 (59.3)Mfold (2) 110 114 67 (60.9) 46 (59.3) 0.599 (60.1)Mfold (3) 110 118 76 (69.1) 37 (67.3) 0.680 (68.2)Sfold (1) 110 116 73 (66.4) 42 (63.5) 0.647 (64.9)Sfold (2) 110 119 86 (78.2) 28 (75.4) 0.767 (76.8)Sfold (3) 110 117 61 (55.5) 55 (52.6) 0.538 (54.0)

Table 4: Note the improvement in prediction accuracy on the supposedly more difficult and longer E.coli RNase P data-set. This shows thatMFE methods are less sensitive to folding errors on longer data-sets but are also less likely to resolve the entire structure. There is littledifference in algorithm accuracy for each of the methods explored here. Each employs the same energy parameters so differences are due toslightly different implementations.

27

Page 28: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Algorithm number of bps number of bps True Positives False Positives Correlationin reference in prediction (% sensitivity) (% selectivity) (%)

E.coli RNase P: Comparative Methods

Plan A: ClustalW Alignment

RNAalifold (H) 71 113 56 (78.9) 16 (77.8) 0.782 (78.3)RNAalifold (H) + RNAfold -C 71 119 55 (77.5) 16 (77.5) 0.773 (77.5)RNAalifold (M) 54 66 31 (57.4) 23 (57.4) 0.571 (57.4)RNAalifold (M) + RNAfold -C 54 77 33 (61.1) 16 (67.3) 0.639 (64.2)Pfold (H) 71 67 47 (66.2) 6 (88.7) 0.765 (77.4)Pfold (M) 54 87 47 (87.0) 4 (92.2) 0.895 (89.6)ILM (H) 71 124 31 (43.7) 54 (36.5) 0.395 (40.1)ILM (M) 54 133 38 (70.4) 31 (55.1) 0.620 (62.7)ILM-pknot (H) 110 124 53 (48.2) 65 (44.9) 0.463 (46.5)ILM-pknot (M) 110 133 44 (40.0) 75 (37.0) 0.382 (38.5)

Plan B: Unaligned sequences

Carnac (H) 71 40 36 (50.7) 0 (100.0) 0.712 (75.4)Carnac (H) + RNAfold -C 71 116 50 (70.4) 25 (66.7) 0.684 (68.5)Carnac (M) 97 80 63 (64.9) 3 (95.5) 0.787 (80.2)Carnac (M) + RNAfold -C 97 118 78 (80.4) 25 (75.7) 0.779 (78.1)Foldalign (H) 71 41 14 (19.7) 25 (35.9) 0.265 (27.8)Foldalign (M) 97 24 5 (5.2) 17 (22.7) 0.107 (13.9)Dynalign (H) 71 95.13 28.63 (40.31) 41.50 (39.59) 0.3974 (39.96)Dynalign (M) 97 103.20 31.00 (31.95) 61.50 (32.80) 0.3208 (32.39)

Plan C: Structure alignment

MARNA (H) 71 89 37 (52.1) 23 (61.7) 0.566 (56.9)MARNA (M) 97 60 48 (49.5) 9 (84.2) 0.645 (66.8)MARNA-trim (H) 71 52 37 (52.1) 3 (92.5) 0.694 (72.3)MARNA-trim (M) 97 43 39 (40.2) 1 (97.5) 0.625 (68.9)

RNAforester (H) 71 114 40 (56.3) 31 (56.3) 0.562 (56.3)RNAforester (M) 97 117 64 (66.0) 44 (59.3) 0.624 (62.6)

Table 5:RNase P is a difficult data-set to study. Five sequences in the high similarity data-set are truncated at both the 5 and 3 prime ends (due tothe primers used for sequencing these). Sequences in the medium similarity data-set are full-length but do not align well using traditionaltools such as clustalw.

28

Page 29: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Algorithm number of bps number of bps True Positives False Positives Correlationin reference in prediction (% sensitivity) (% selectivity) (%)

E.coli SSU rRNA: Single Sequence Methods

RNAfold 468 493 207 (44.2) 271 (43.3) 0.437 (43.8)Mfold (1) 468 480 240 (51.3) 224 (51.7) 0.515 (51.5)Mfold (2) 468 487 242 (51.7) 229 (51.4) 0.515 (51.5)Mfold (3) 468 487 202 (43.2) 273 (42.5) 0.428 (42.8)Sfold (1) 468 481 232 (49.6) 229 (50.3) 0.499 (49.9)Sfold (2) 468 499 231 (49.4) 249 (48.1) 0.487 (48.7)Sfold (3) 468 475 232 (49.6) 230 (50.2) 0.498 (49.9)

Table 6:

29

Page 30: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Algorithm number of bps number of bps True Positives False Positives Correlationin reference in prediction (% sensitivity) (% selectivity) (%)

E.coli SSU rRNA: Comparative Methods

Plan A: ClustalW Alignment

RNAalifold (H) 460 472 275 (59.8) 179 (60.6) 0.601 (60.2)RNAalifold (H) + RNAfold -C 460 483 273 (59.3) 195 (58.3) 0.588 (58.8)RNAalifold (M) 441 433 372 (84.4) 32 (92.1) 0.881 (88.2)RNAalifold (M) + RNAfold -C 441 469 388 (88.0) 44 (89.8) 0.889 (88.9)Pfold (H) 460 377 326 (70.9) 26 (92.6) 0.810 (81.7)Pfold (M) 441 0 0 (0.0) 0 (0.0) 0.000 (0.0)ILM (H) 460 565 236 (51.3) 313 (43.0) 0.469 (47.1)ILM (M) 441 564 264 (59.9) 249 (51.5) 0.554 (55.7)ILM-pknot (H) 468 565 236 (50.4) 311 (43.1) 0.466 (46.8)ILM-pknot (M) 468 564 266 (56.8) 258 (50.8) 0.537 (53.8)

Plan B: Unaligned sequences

Carnac (H) 460 233 206 (44.8) 12 (94.5) 0.650 (69.6)Carnac (H) + RNAfold -C 460 470 332 (72.2) 112 (74.8) 0.734 (73.5)Carnac (M) 448 294 259 (57.8) 18 (93.5) 0.735 (75.7)Carnac (M) + RNAfold -C 448 471 337 (75.2) 110 (75.4) 0.753 (75.3)

Table 7:The probabilistic approach of PFold can, on occasion, suffer from “under-flow” errors caused by multiplying many probabilities togetherproducing numbers too low to be dealt with on modern computers. This is what has happened on the medium similarity data-set.

30

Page 31: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Algorithm number of bps number of bps True Positives False Positives Correlationin reference in prediction (% sensitivity) (% selectivity) (%)

E.coli LSU rRNA: Single Sequence Methods

RNAfold 839 906 435 (51.8) 430 (50.3) 0.510 (51.1)Mfold (1) 839 883 458 (54.6) 383 (54.5) 0.545 (54.5)Mfold (2) 839 892 480 (57.2) 364 (56.9) 0.570 (57.0)Mfold (3) 839 889 454 (54.1) 392 (53.7) 0.539 (53.9)Sfold (1) 839 903 49 (5.8) 811 (5.7) 0.057 (5.8)Sfold (2) 839 878 432 (51.5) 411 (51.2) 0.513 (51.4)Sfold (3) 839 882 384 (45.8) 463 (45.3) 0.455 (45.6)

Table 8:

31

Page 32: A comprehensive comparison of comparative RNA structure ...projects.binf.ku.dk/pgardner/bralibase/GardnerGiegerich0...A comprehensive comparison of comparative RNA structure prediction

Algorithm number of bps number of bps True Positives False Positives Correlationin reference in prediction (% sensitivity) (% selectivity) (%)

E.coli LSU rRNA: Comparative Methods

Plan A: ClustalW Alignment

RNAalifold (H) 794 879 627 (79.0) 195 (76.3) 0.776 (77.6)RNAalifold (H) + RNAfold -C 794 871 629 (79.2) 185 (77.3) 0.782 (78.2)RNAalifold (M) 819 721 614 (75.0) 53 (92.1) 0.831 (83.5)RNAalifold (M) + RNAfold -C 819 790 691 (84.4) 78 (89.9) 0.871 (87.1)Pfold (H) 794 0 0 (0.0) 0 (0.0) 0.000 (0.0)Pfold (M) 819 0 0 (0.0) 0 (0.0) 0.000 (0.0)ILM (H) 794 1048 389 (49.0) 602 (39.3) 0.438 (44.1)ILM (M) 819 1161 560 (68.4) 405 (58.0) 0.630 (63.2)ILM-pknot (H) 869 1048 272 (31.3) 759 (26.4) 0.287 (28.8)ILM-pknot (M) 869 1161 377 (43.4) 629 (37.5) 0.403 (40.4)

Plan B: Unaligned sequences

Carnac (H) 816 422 390 (47.8) 7 (98.2) 0.685 (73.0)Carnac (H) + RNAfold -C 816 873 674 (82.6) 156 (81.2) 0.819 (81.9)Carnac (M) 821 508 463 (56.4) 14 (97.1) 0.740 (76.7)Carnac (M) + RNAfold -C 821 865 682 (83.1) 147 (82.3) 0.827 (82.7)

Table 9:

32